Nihms 630759

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 20

NIH Public Access

Author Manuscript
Vet Clin North Am Small Anim Pract. Author manuscript; available in PMC 2015 November 01.
Published in final edited form as:
NIH-PA Author Manuscript

Vet Clin North Am Small Anim Pract. 2014 November ; 44(6): 1113–1129. doi:10.1016/j.cvsm.
2014.07.008.

Aging in the Canine and Feline Brain


Charles H. Vite, DVM, PhDa and Elizabeth Head, MA, PhDb,*
aDepartment of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania,
Section of Neurology & Neurosurgery, Department of Clinical Studies - Philadelphia, 3900
Delancey Street, Philadelphia, PA 19104, USA
bDepartment of Pharmacology & Nutritional Sciences, Sanders-Brown Center on Aging,
University of Kentucky, 800 South Limestone Street, 203 Sanders Brown Building, Lexington, KY
40515, USA

Keywords
NIH-PA Author Manuscript

β-Amyloid; Cat; Dog; Cognitive dysfunction; Neuron loss; Tau

INTRODUCTION
This article reviews canine and feline brain aging. Several key features are discussed and
compared, including general aging characteristics and neuropathology. Aging dogs and cats
show many similarities in terms of brain changes but also some important differences.
Several research groups have been working with aging dogs and cats to test various theories
of aging and to develop therapeutics that will be beneficial to both species.

The median life span of dogs varies as a function of breed, with larger breeds typically
having shorter life spans than smaller breeds.1–3 For the purposes of this article, several
studies that are described have been collected in purpose-bred beagles and additional
companion animals and clinical data are shared when available. Beagles have a median life
span of 13.9 years, with no significant differences between males and females.4 A young
NIH-PA Author Manuscript

beagle less than 5 years old is similar to humans who are less than 40 years old.3 Middle-
aged beagles between 5 and 9 years are similar to humans between 40 and 60 years and
beagles more than 9 years old are similar to humans more than 60 years old. However, the
larger the breed of dog, the shorter the life span and thus biological age may vary across
breeds given a specific age.1

In a laboratory setting and in the veterinary clinic, studies of aging dogs report that some but
not all aged dogs are impaired on different measures of learning and memory (see Refs.5–7).
Not all old dogs are impaired and not all types of learning and memory are equally affected.
Neurobiological changes, as described later, can account for some, but not all, of the clinical
signs of cognitive decline in aging dogs.

© 2014 Elsevier Inc. All rights reserved.


*
Corresponding author. elizabeth.head@uky.edu.
Vite and Head Page 2

NEUROBIOLOGY OF AGING IN THE DOG


This article describes several neurobiological changes associated with aging in dogs (Table
NIH-PA Author Manuscript

1).

Brain Atrophy
Old dogs often show marked ventriculomegaly at postmortem examination associated with
thinning of the cerebral cortex and the subcortical white matter.8 Magnetic resonance
imaging (MRI) studies performed on aged beagle and German shepherd dogs show that
cortical atrophy, identified as widened sulci, thinned parenchyma,9 and ventricular
dilatation,9–11 progresses with age and is a consistent feature of canine brain aging.
Furthermore, MRI studies suggest differential vulnerabilities of specific brain areas to aging.
For example, in aging beagle dogs, the prefrontal cortex loses tissue volume at an earlier age
(approximately 8–11 years) than does the hippocampus (after 11 years).12 An MRI study by
Hasegawa and colleagues13 (2005) suggested that although interthalamic adhesion thickness
was smaller in older dogs, those showing age-related cognitive decline and a single dog with
GM1 gangliosidosis also had a significant decrease in the thickness.
NIH-PA Author Manuscript

Periventricular white matter signal abnormalities are frequently seen in MRI of various dog
breeds greater than 12 years of age that are evaluated for seizures, vestibular disease, or
behavioral abnormalities. These bilaterally symmetric T2-weighted hyperintensities of the
internal capsule are suspected to be caused by wallerian degeneration, demyelination, and
accompanying gliosis.14

Selective Neuron Loss


Atrophy may result from neuron loss or changes in neuronal density, as reported in normal
human brain aging,15,16 although more extensive neuronal loss occurs in Alzheimer disease
(AD).17,18 When neurons were counted using unbiased stereological methods within
individual subfields of the hippocampus of young (3.4–4.5 years) and old (13.0–15.0 years)
dogs, the aged dogs had significantly (~30%) fewer neurons in the hilus of the dentate
gyrus.19 A study by Pugliese and colleagues20 (2007) showed that cognitive deficits
correlated with loss of Purkinje cells in the cerebellum. More recently, a study by Insua and
colleagues21 (2010) examined noradrenergic neurons in the locus coeruleus of aged canines,
a group of neurons that are implicated in AD in humans.22,23 Dogs that are cognitively
NIH-PA Author Manuscript

impaired show a significant reduction in noradrenergic neurons. Reduced neurogenesis in


the mature brain may also contribute to reduced neuron numbers and age-associated
cognitive decline, resulting in slower replacement of dying neurons. In the hippocampus of
beagles, a 90% to 95% decline in neurogenesis was measured in aged dogs.24 Similar
reductions in neurogenesis in aged dogs have been reported by other investigators.25

Senile Plaques (β-Amyloid) in Dogs


A key feature of canine brain aging was the observation in 1956, by Braumühl, who reported
Alzheimer-like senile plaques in aged dogs (reviewed in Ref.26). Senile plaque accumulation
in the aged canine brain has been well described.26 Senile plaques are composed of β-
amyloid (Aβ) protein, and are one of 2 key types of neurologic pathologies observed in the

Vet Clin North Am Small Anim Pract. Author manuscript; available in PMC 2015 November 01.
Vite and Head Page 3

AD brain.27 The Aβ peptide is produced by the sequential cleavage of the amyloid precursor
protein (APP) by beta-secretase and gamma-secretase.28,29 Cleavage by gamma-secretase
results in differing lengths of Aβ, with the 42 amino acid form, Aβ1-42 making up most of
NIH-PA Author Manuscript

the insoluble deposits found in the AD brain.30 One of the reasons why the canine brain has
been examined extensively for Aβ neurologic disorder is that dogs and humans share an
identical amino acid sequence of the protein.31,32 Aβ is thought to be a causative factor for
AD in people.33 The observation of brain Aβ first stimulated interest in the use of the dog to
model human aging and disease.34 Diffuse plaques are the predominant subtype of Aβ in the
aging dog brain (Fig. 1).35–40 Specific brain regions are differentially vulnerable to
Aβ.36,41–46 When cortical regions are sampled for Aβ deposition, each region shows a
different age of Aβ onset.43 In the dog, Aβ deposition occurs earliest in the prefrontal cortex
(see Fig. 1C, D) and later in the temporal cortex, hippocampus (see Fig. 1A, B), and
occipital cortex.45

Beagles that show learning and memory impairments in a laboratory setting with systematic
cognitive tests also show higher levels of Aβ plaques than those old dogs without cognitive
impairments.47–50 For example, dogs with prefrontal cortex–dependent reversal learning
deficits show significantly higher amounts of Aβ in this brain region.48,49 As in laboratory
NIH-PA Author Manuscript

beagles, the extent of Aβ plaques varies as a function of age in companion dogs (including a
wide variety of breeds and mixed breeds).50–52 Further, the extent of Aβ plaques correlates
with behavior changes and this association remains significant even if age is removed as a
covariate.47,50 Case report 1 and Figs. 1 and 2 show a case study of cognitive decline and
age-associated Aβ neurologic disorder in a border collie.

Case report 1
Martha was a female border collie that was a very competent farm dog with what her
owner called “lots of sheep savvy.” She was also a fully integrated family member and
companion to the family when hiking, skiing, and mountain biking. In addition, she
successfully competed in sheep dog trials till the age of 10 years (see Fig. 1A). She and
her handler worked their way through the succession of novice-level trials, winning
several, and ultimately competing and placing in the highest level, the Open class, which
attests to her skill (see Fig. 1B).

By the time Martha was 11 years old she was nearly deaf and slow enough that she could
NIH-PA Author Manuscript

no longer trial. She started to show some confusion with commands at which she was
expert. For example, one day her owner took her out in the field and gave her a command
to go counter clockwise around the sheep and she went clockwise. Further, when her
owner next said “OK, go clockwise” she reversed her direction, when she should have
continued the clockwise direction (see Fig. 1C). The next time her owner was working
with her, a normally benign ewe turned around to face Martha and stomped her foot.
Martha, who would normally have politely stood her ground until the sheep backed
down, promptly turned away and left the field. The owner at this time thought, “Well that
is proof that Martha is fully retired now.” Not long after the incident with the ewe, the
sheep began to act as if Martha did not even exist.

Vet Clin North Am Small Anim Pract. Author manuscript; available in PMC 2015 November 01.
Vite and Head Page 4

Over the next 2 years, changes in Martha were subtle and included increasing hearing
loss and stubbornness. She also stopped having interest in the sheep, and appeared most
fixated with another dog (Ida) in the family. Several behaviors with the other dog
NIH-PA Author Manuscript

appeared abnormal. For example, Martha would stare at Ida, the middle dog, whether
they were inside or out walking, and then would stalk Ida, despite a lack of interest from
Ida in interacting. The owners had to intervene frequently to prevent a snarling episode,
although the two dogs never injured each other.

To her last day, Martha’s social skills with human friends persisted. However, she began
pacing, panting, and getting stuck under the bedside table at night. During the day when
only 1 of the owners was home, Martha was frequently anxious and sometimes clingy,
which never used to be the case. She showed signs of disorientation and during walks
occasionally lost the younger dogs and her owner although the same daily route was
followed. Martha was once found in a road looking up and down as if lost, although she
had been walking that direction for years.

Throughout her life Martha had a great appetite and generally excellent health, although
she had arthritis in almost every joint in her body and was on high doses of incontinence
medication and Rimadyl (a nonsteroidal antiinflammatory drug) for arthritis. For the last
NIH-PA Author Manuscript

year of her life, the owner was unable to get a photograph of Martha looking at her
because she was no longer making eye contact (see Fig. 1D).

When Martha was 14 years old, the decision was made to euthanize her and the owner
donated her brain to the University of Kentucky to determine whether neurologic
disorder was present. Aβ neurologic disorder was extensive in Martha (see Fig. 2).

The focus in AD pathogenesis has recently shifted from Aβ plaques to considering smaller,
soluble forms of Aβ assemblies called Aβ oligomers. Oligomers are highly toxic and impair
synaptic function.53 Furthermore, increased oligomer levels are strongly associated with
cognitive dysfunction.53–55 A recent study by Pop and colleagues,56 examined the
accumulation of oligomeric Aβ in the temporal lobe of canines. This study provided
evidence that canines, like humans, experience an increase in toxic oligomers with age.

Vascular Disorders, Cerebrovascular Amyloid Angiopathy


NIH-PA Author Manuscript

Cerebrovascular amyloid angiopathy (CAA) is the deposition of Aβ in association with the


cerebrovasculature (see Fig. 1D). In dogs with CAA, the blood vessels of the brain typically
contain the shorter, 40 amino acid–long species of Aβ.57–59 The occipital cortex seems to be
particularly vulnerable to CAA in the aged dog brain. Vascular Aβ may compromise the
blood-brain barrier, disrupt vessel wall viability,60 and cause microhemorrhages.61

Aged dogs may also show lacunar infarcts of the caudate nucleus and thalamus with most
dogs showing no causative metabolic, endocrine, or hypertensive disease.62,63 In a
longitudinal study, these lesions were shown to increase in number with advancing age.64
Lacunar infarcts of the caudate nucleus have been induced experimentally in beagle dogs by
proximal middle cerebral artery occlusion.65 The cause of naturally occurring lacunar
infarcts in dogs remains unidentified.

Vet Clin North Am Small Anim Pract. Author manuscript; available in PMC 2015 November 01.
Vite and Head Page 5

Tau Neuropathology
Unlike humans but like many other animal species, canines do not develop full-blown
neurofibrillary tangles,32,37,39,40 which is the second neuropathologic characteristic of AD.
NIH-PA Author Manuscript

However, aged dogs show the accumulation of several phosphorylated tau epitopes that are
consistent with AD in humans and observations in aged cats (described later).66 In a study of
cognitively impaired pet dogs that included a variety of breeds, intracellular phosphorylated
tau was observed that was similar to the AD brain.67

Oxidative Damage and Mitochondrial Dysfunction


Aging and the production of free radicals can lead to oxidative damage to proteins, lipids,
and nucleotides that, in turn, may cause neuronal dysfunction and ultimately neuronal death.
In the aging dog, the brain accumulates carbonyl groups, which are a measure of oxidative
damage to proteins.68,69 Carbonyl groups are associated with reduced endogenous
antioxidant enzyme activity or protein levels, including those of glutamine synthetase and
superoxide dismutase (SOD).68,70–72 In addition, increased oxidative damage to proteins can
be measured by the end products of lipid peroxidation (oxidative damage to lipids),
including 4-hydroxynonenal,50,52,72,73 lipofuscin,50 lipofuscinlike pigments,52,73 or
malondialdehyde.68 In addition, oxidative damage to DNA or RNA may be increased in the
NIH-PA Author Manuscript

aged dog brain.5,50

Oxidative damage may also be associated with behavioral decline in dogs. Increased
oxidative end products in the aged companion dog brain are correlated with more severe
behavioral changes.50,52,69 In laboratory studies of aging beagles, higher protein oxidative
damage (3-nitrotyrosine) and lower endogenous antioxidant capacity (SOD and glutathione-
S-transferase activities) are associated with poorer prefrontal-dependent and spatial
learning.71 Mitochondria are sources of free radicals that damage proteins, lipids, and DNA/
RNA.74 In a study of aged beagles, isolated mitochondria showed increased reactive oxygen
species production in aged animals relative to young animals.75 Thus, aged dogs show
mitochondrial dysfunction and oxidative damage, consistent with humans with age-related
neurologic dysfunction.

Correlates of Brain Aging Found in Lysosomal Storage Diseases


Cross-sectional studies of brain aging are negatively influenced by interindividual variations
NIH-PA Author Manuscript

in brain size and structure, as well as by differing rates of atrophy. Longitudinal studies may
be limited by repeated access to aged animals and to subject attrition.64 Insight into
mechanisms of brain aging can also be obtained through the study of breeding colonies of
dogs with naturally occurring hereditary metabolic disorders, including mitochondrial
disorders, lysosomal storage diseases, and leukodystrophies, which have clinical,
neuropathologic, and biochemical changes similar to those found in old dogs. These
disorders allow both cross-sectional and longitudinal studies of disease progression in
related animals with short life spans.76,77 For example, more than 40 inherited lysosomal
storage diseases (LSDs) have been identified, with many characterized by progressive
cognitive decline, memory loss, brain atrophy, loss of myelin, and region-specific loss of
neurons as are seen in aging.78,79 Two well-studied diseases are examples of what has been
learned by studying affected dogs. The neuronal ceroid lipofuscinoses (NCLs) are

Vet Clin North Am Small Anim Pract. Author manuscript; available in PMC 2015 November 01.
Vite and Head Page 6

characterized by seizures, motor dysfunction, impaired vision, progressive cognitive decline,


impaired memory, behavioral problems, brain degeneration, selective neuronal loss that is
most severe in the cerebral cortex, white matter atrophy, neuronal accumulation of protein
NIH-PA Author Manuscript

and lipid, and premature death.80,81 Both human patients and affected dogs show various
ages of onset, disease course, and neuropathology.80–82 The juvenile-onset form of the
disease, known as Batten disease, has naturally occurring analogous disease in English
setters, Tibetan terriers, and border collies.83 Biochemical analysis of the brain shows that
storage of subunit c of the mitochondrial ATP synthase complex occurs; subunit c is an
essential membrane component of the proton channel of the ATP synthase complex, which
generates ATP by oxidative phosphorylation.81 Although not fully characterized, the
neuronal loss seen in Batten disease is postulated to be caused by mitochondrial dysfunction
and energy-linked excitotoxicity.83 Synaptic loss and glial activation also occur in this
disease, although the mechanism and contribution of these abnormalities to disease
progression are not yet understood.80

The T-maze reversal learning task has been used to evaluate progressive cognitive decline in
the dachshund model of late-infantile NCL, which is caused by a mutation in the gene
encoding the enzyme tripeptidyl-peptidase 1 (TTP1).84 This model system is particularly
NIH-PA Author Manuscript

interesting because intrathecal administration of TTP1 results in decreased lysosomal


storage.85 It is likely that a better understanding of how mitochondrial dysfunction and
neuronal loss develop in the NCLs, as well as how they may be reversed with therapy, will
shed light on similar mechanisms postulated to occur in brain aging.

Mucopolysaccharidosis type I (MPSI; Hurler syndrome) is another LSD characterized by


progressive cognitive decline, including impaired memory and intelligence, in human
patients.86 Affected humans and dogs show progressive cortical atrophy, ventricular
enlargement, and white matter loss.87 Cardiovascular disease and accumulation of
intracellular Aβ are also described in affected patients.88,89 In human patients with MPSI,
MRI showed that corpus callosum volume and fractional anisotropy correlated with
neuropsychological testing.86 In dogs with MPSI, imaging studies showed corpus callosum
volumes to be smaller in affected dogs compared with unaffected dogs; no cognitive studies
have yet been performed. Similar to the late-infantile NCL, this disease is amenable to either
whole-brain therapy by intrathecal enzyme replacement therapy or to regional gene therapy
allowing for the assessment of therapeutic effect.90,91 When treated with intrathecal alpha-
NIH-PA Author Manuscript

iduronidase, the enzyme that is deficient in MPSI, corpus callosum volumes in affected dogs
were no longer distinguishable from those in unaffected dogs. These studies indicate the
usefulness of quantitative imaging to study brain atrophy over time as well as the
effectiveness of therapy in ameliorating the atrophy.

NEUROBIOLOGY OF AGING IN THE CAT


Cats are considered to be old, or senior, starting around the age of 7 to 10 years but
consistently after 12 years, depending on the individual animal. Aging cats show several
behavioral changes that can be of concern to a pet owner and that are not related to systemic
illness or disease.92 Whether there is cognitive decline in aging cats as observed in dogs is
still being studied but there are behavioral changes that have been reported clinically.93

Vet Clin North Am Small Anim Pract. Author manuscript; available in PMC 2015 November 01.
Vite and Head Page 7

There are fewer studies of the aging cat brain compared with aging dogs in the literature,
highlighting the significant gaps in knowledge regarding feline brain aging. Several reviews
have been written describing feline age-associated neurologic disorder and some of those
NIH-PA Author Manuscript

observations are summarized here.92,93

Neuron Loss and Atrophy


There are several studies suggesting that the caudate nucleus of aging cats is affected by
aging, including reduced neuronal numbers and reduction in the density of synapses.94–97
These losses may lead to impairments in motor function.94 The locus coeruleus, a key
nucleus responsible for producing the neurotransmitter acetylcholine, which is associated
with learning and memory, also shows neuronal losses with age in cats.98


Aβ is typically observed in cats more than the age of 10 years,99–103 although there is a
report of 7.5-year-old animal showing Aβ disorder.102 Feline Aβ deposition seems to be
different from that of dogs and humans in several respects. Unlike human and dog brain,
plaques in the cat brain are primarily made up of Aβ1-42 without Aβ1-40 and the peptide is
not posttranslationally modified (suggesting a more rapid turnover) in cats compared with
NIH-PA Author Manuscript

dogs and humans.66,99 Further, truncated Aβ (AβpN3) was absent in aged cat brain.104
Blood vessels in aging cat brains are positive for Aβ1-40, the shorter more soluble form of
Aβ.66 To our knowledge, there are few reports of the link between behavioral dysfunction
and the extent of Aβ disorder as reported in dogs. Although cats that show signs of
behavioral dysfunction tend to also have Aβ plaques,100 the severity of behavioral changes
does not seem to correlate well with the extent of Aβ disorder.66

Tau Phosphorylation
An interesting feature of the aging cat brain is the detection of phosphorylated tau protein,
which is consistent with reports in human AD.105,106 Cats show multiple isoforms of tau
protein, as do humans.107 Not all studies consistently observe tau neuropathology in
cats101,108,109 and this may be because of methodological challenges. In addition, when
using sensitive immunohistochemical methods, phosphorylated tau is not as frequently
observed as Aβ plaques, but the epitopes on tau that are phosphorylated overlap with those
observed in dogs and humans.66 The morphology of neurons that show an accumulation of
NIH-PA Author Manuscript

intracellular phosphorylated tau suggests a sprouting response, which is also similar to


human brain.66 Tau phosphorylation is also more frequently associated with the presence of
seizures in aging cats.100

Neuronal Loss in Feline Niemann-Pick Type C Disease and Similarities to AD


Niemann-Pick type C (NPC) disease is another example of how LSDs may contribute to the
understanding of neuronal loss, oxidative stress, Aβ deposition, and tau neuropathology in
the aging brain. NPC disease is caused by dysfunction of either of 2 proteins, NPC1 or
NPC2, which result in lysosomal storage of cholesterol and glycosphingolipids.110 How
dysfunction of these proteins results in the dementia and neuronal loss associated with
disease has been difficult to determine. A feline model of NPC1 disease exists and has been

Vet Clin North Am Small Anim Pract. Author manuscript; available in PMC 2015 November 01.
Vite and Head Page 8

critical for understanding disease pathogenesis and evaluating therapy.110–115 As disease-


modifying therapies are evaluated, clinicians hope to gain insight into the relative
contributions of each of the following factors in causing cognitive decline and brain atrophy.
NIH-PA Author Manuscript

Autophagy, the degradation or recycling of damaged intracellular organelles and


aggregated-proteins, is necessary for cellular homeostasis. NPC1 has been implicated in
mediating membrane-tethering events between autophagosomes and late endosomes that
subsequently fuse with lysosomes to degrade their contents.116 When NPC1 is defective,
autophagy is impaired. Because impaired basal autophagy has been found to cause
neurodegeneration,117,118 it is postulated that impaired autophagy in NPC1 disease may
contribute to the observed neuronal loss as a consequence of the buildup of dysfunctional
mitochondria and the accumulation of misfolded proteins, resulting in cell death. Altered
autophagosomal-lysosomal function has similarly been implicated in brain aging.119,120

Oxidative stress and the generation of reactive oxygen species have also been proposed as
contributing factors for neuronal loss in NPC disease.121 In vitro studies showed increased
oxidative stress in cultured neurons and fibroblasts.122 Evaluation of serum from patients
with NPC showed decreased fractions of reduced coenzyme Q10 and decreased Trolox-
equivalent antioxidant capacity.121 Evaluation of plasma and cerebrospinal fluid from
NIH-PA Author Manuscript

patients and affected cats showed accumulation of cholesterol oxidation products, and, in
cats, these oxysterols decreased in response to a disease-modifying therapy.122

In addition, both NPC1 disease and AD are marked by dementia and abnormal cholesterol
metabolism; however, they also share abnormalities in Aβ processing and the presence of
neurofibrillary tangles.110,123 First, in both affected humans and cats, relative Aβ peptide
distributions differ from those found in unaffected individuals, with lower relative levels of
Aβ1-37, Aβ1-38 and Aβ1-39 in cats with NPC1 compared with controls.124 Second,
neurofibrillary tangles composed of paired helical filaments of phosphorylated microtubular
protein accumulate in NPC disease as they do in AD.110 Third, apolipoprotein E4, the
isoform associated with increased risk for developing AD, has also been found to be
associated with early onset of disease and with increased NPC1 disease severity.123 Other
deficits seen in NPC1 disease, including peroxisomal dysfunction,125 abnormal sphingosine
metabolism,126 neuroinflammation,127 and induction of apoptosis,128 may also contribute to
changes seen in the aging brain.129
NIH-PA Author Manuscript

CLINICAL IMPLICATIONS OF BRAIN AGING IN DOGS AND CATS


Laboratory-based studies of cognition in beagles suggest that there are age-dependent
functional changes related to brain disorders. In the clinic, owners of geriatric dogs
frequently report behavioral changes.130–132 Some of these behavioral changes may be
linked to systemic illness or other health issues (including sensory decline). When ruled out,
a subset of older dogs shows evidence of behavioral changes that are now considered to be
signs of canine cognitive dysfunction (CCD). The original reports of CCD were by Ruehl
and colleagues133 in the mid 1990s and several reports followed, along with the
development of new tools to detect CCD in pet dogs.6,7,93,134,135 CCD has been linked to
brain pathology in pet dogs.20,136,137 There are reports that CCD can be reduced in aging

Vet Clin North Am Small Anim Pract. Author manuscript; available in PMC 2015 November 01.
Vite and Head Page 9

dogs through treatment with various pharmaceuticals (eg, Anipryl),134 by dietary


intervention using a prescription diet (B/D diet Hills Pet Nutrition134; the same diet used in
laboratory studies showing a benefit to cognition and neuropathology138), and to
NIH-PA Author Manuscript

immunotherapy using anti-Aβ approaches.139 In cats, there is less evidence of a form of


feline cognitive dysfunction syndrome but evidence is accumulating that a similar
phenomenon can occur.66,92,93,100,140

SUMMARY
Aging dogs and cats show features of brain disorders that can be similar to human aging and
AD. Neuropathologic changes with age may be linked to signs of cognitive dysfunction both
in the laboratory and in a clinic setting. Less is known about cat brain aging and cognition
and this represents an area for further study. Neurodegenerative diseases such as LSDs in
dogs and cats also show similar features of aging, suggesting some common underlying
pathogenic mechanisms and also suggesting pathways that can be modified to promote
healthy brain aging.

Acknowledgments
NIH-PA Author Manuscript

The authors are grateful to Virginia Price, DVM, and her stories of Martha and donation of her tissue for research.
We thank Judianne Davis-Van Nostrand and Dr William Van Nostrand at Stony Brook University in New York for
help with collecting tissue for the study. Ms Katie McCarty collected the Aβ immunostaining data for Martha.

Research reported in this publication was supported by the National Institute on Aging of the National Institutes of
Health under award number R01AG031764 (E. Head) and R01NS073661 and P40-02512 (C.H. Vite, Mark
Haskins), and the Ara Parseghian Medical Research Foundation (C.H. Vite). The content is solely the responsibility
of the authors and does not necessarily represent the official views of the National Institutes of Health.

References
1. Greer KA, Canterberry SC, Murphy KE. Statistical analysis regarding the effects of height and
weight on life span of the domestic dog. Res Vet Sci. 2007; 82(2):208–14. [PubMed: 16919689]
2. Galis F, Van der Sluijs I, Van Dooren TJ, et al. Do large dogs die young? J Exp Zool B Mol Dev
Evol. 2007; 308(2):119–26. [PubMed: 16788896]
3. Patronek GJ, Waters DJ, Glickman LT. Comparative longevity of pet dogs and humans:
implications for gerontology research. J Gerontol A Biol Sci Med Sci. 1997; 52(3):B171–8.
[PubMed: 9158552]
4. Lowseth LA, Gillett NA, Gerlach RF, et al. The effects of aging on hematology and serum
NIH-PA Author Manuscript

chemistry values in the beagle dog. Vet Clin Pathol. 1990; 19(1):13–9. [PubMed: 12684941]
5. Cotman CW, Head E. The canine (dog) model of human aging and disease: dietary, environmental
and immunotherapy approaches. J Alzheimers Dis. 2008; 15(4):685–707. [PubMed: 19096165]
6. Landsberg G, Araujo JA. Behavior problems in geriatric pets. Vet Clin North Am Small Anim
Pract. 2005; 35(3):675–98. [PubMed: 15833565]
7. Landsberg G, Ruehl W. Geriatric behavioral problems. Vet Clin North Am Small Anim Pract. 1997;
27(6):1537–59. [PubMed: 9348642]
8. Vandevelde, M.; Higgins, RJ.; Oevermann, A. Veterinary neuropathology: essentials of theory and
practice. West Sussex (United Kingdom): Wiley-Blackwell; 2012.
9. Su MY, Head E, Brooks WM, et al. MR imaging of anatomic and vascular characteristics in a
canine model of human aging. Neurobiol Aging. 1998; 19(5):479–85. [PubMed: 9880050]
10. Kimotsuki T, Nagaoka T, Yasuda M, et al. Changes of magnetic resonance imaging on the brain in
beagle dogs with aging. J Vet Med Sci. 2005; 67(10):961–7. [PubMed: 16276050]

Vet Clin North Am Small Anim Pract. Author manuscript; available in PMC 2015 November 01.
Vite and Head Page 10

11. Gonzalez-Soriano J, Marin Garcia P, Contreras-Rodriguez J, et al. Age-related changes in the


ventricular system of the dog brain. Ann Anat. 2001; 183(3):283–91. [PubMed: 11396800]
12. Tapp PD, Siwak CT, Gao FQ, et al. Frontal lobe volume, function, and beta-amyloid pathology in
NIH-PA Author Manuscript

a canine model of aging. J Neurosci. 2004; 24(38):8205–13. [PubMed: 15385603]


13. Hasegawa D, Yayoshi N, Fujita Y, et al. Measurement of interthalamic adhesion thickness as a
criteria for brain atrophy in dogs with and without cognitive dysfunction (dementia). Vet Radiol
Ultrasound. 2005; 46(6):452–7. [PubMed: 16396259]
14. Grossman, RI.; Yousem, DM. Neuroradiology: the requisites. Philadelphia: Mosby; 2003.
15. Simic G, Kostovic I, Winblad B, et al. Volume and number of neurons of the human hippocampal
formation in normal aging and Alzheimer’s disease. J Comp Neurol. 1997; 379(4):482–94.
[PubMed: 9067838]
16. West MJ. Regionally specific loss of neurons in the aging human hippocampus. Neurobiol Aging.
1993; 14:287–93. [PubMed: 8367010]
17. West MJ, Kawas CH, Martin LJ, et al. The CA1 region of the human hippocampus is a hot spot in
Alzheimer’s disease. Ann N Y Acad Sci. 2000; 908:255–9. [PubMed: 10911964]
18. Bobinski M, Wegiel J, Tarnawski M, et al. Relationships between regional neuronal loss and
neurofibrillary changes in the hippocampal formation and duration and severity of Alzheimer
disease. J Neuropathol Exp Neurol. 1997; 56(4):414–20. [PubMed: 9100672]
19. Siwak-Tapp CT, Head E, Muggenburg BA, et al. Region specific neuron loss in the aged canine
hippocampus is reduced by enrichment. Neurobiol Aging. 2008; 29(1):39–50. [PubMed:
17092609]
NIH-PA Author Manuscript

20. Pugliese M, Gangitano C, Ceccariglia S, et al. Canine cognitive dysfunction and the cerebellum:
acetylcholinesterase reduction, neuronal and glial changes. Brain Res. 2007; 1139:85–94.
[PubMed: 17292335]
21. Insua D, Suarez ML, Santamarina G, et al. Dogs with canine counterpart of Alzheimer’s disease
lose noradrenergic neurons. Neurobiol Aging. 2008; 31(4):625–35. [PubMed: 18573571]
22. Dringenberg HC. Alzheimer’s disease: more than a ‘cholinergic disorder’ - evidence that
cholinergic-monoaminergic interactions contribute to EEG slowing and dementia. Behav Brain
Res. 2000; 115(2):235–49. [PubMed: 11000423]
23. Grudzien A, Shaw P, Weintraub S, et al. Locus coeruleus neurofibrillary degeneration in aging,
mild cognitive impairment and early Alzheimer’s disease. Neurobiol Aging. 2007; 28(3):327–35.
[PubMed: 16574280]
24. Siwak-Tapp CT, Head E, Muggenburg BA, et al. Neurogenesis decreases with age in the canine
hippocampus and correlates with cognitive function. Neurobiol Learn Mem. 2007; 88(2):249–59.
[PubMed: 17587610]
25. Pekcec A, Baumgartner W, Bankstahl JP, et al. Effect of aging on neurogenesis in the canine brain.
Aging Cell. 2008; 7(3):368–74. [PubMed: 18363905]
26. Cummings BJ, Head E, Ruehl W, et al. The canine as an animal model of human aging and
dementia. Neurobiol Aging. 1996; 17(2):259–68. [PubMed: 8744407]
NIH-PA Author Manuscript

27. Mirra SS, Heyman A, McKeel D, et al. The Consortium to Establish a Registry for Alzheimer’s
Disease (CERAD). Part II. Standardization of the neuropathologic assessment of Alzheimer’s
disease. Neurology. 1991; 41(4):479–86. [PubMed: 2011243]
28. Murphy MP, LeVine H 3rd. Alzheimer’s disease and the amyloid-beta peptide. J Alzheimers Dis.
2010; 19(1):311–23. [PubMed: 20061647]
29. Selkoe DJ. Amyloid beta-protein and the genetics of Alzheimer’s disease. J Biol Chem. 1996;
271:18295–8. [PubMed: 8756120]
30. Selkoe DJ. Alzheimer’s disease: genes, proteins, and therapy. Physiol Rev. 2001; 81(2):741–66.
[PubMed: 11274343]
31. Johnstone EM, Chaney MO, Norris FH, et al. Conservation of the sequence of the Alzheimer’s
disease amyloid peptide in dog, polar bear and five other mammals by cross-species polymerase
chain reaction analysis. Brain Res Mol Brain Res. 1991; 10(4):299–305. [PubMed: 1656157]
32. Selkoe DJ, Bell DS, Podlisny MB, et al. Conservation of brain amyloid proteins in aged mammals
and humans with Alzheimer’s disease. Science. 1987; 235(4791):873–7. [PubMed: 3544219]

Vet Clin North Am Small Anim Pract. Author manuscript; available in PMC 2015 November 01.
Vite and Head Page 11

33. Hardy J. Alzheimer’s disease: the amyloid cascade hypothesis: an update and reappraisal. J
Alzheimers Dis. 2006; 9(3 Suppl):151–3. [PubMed: 16914853]
34. Wisniewski, HM.; Wegiel, J.; Morys, J., et al. Aged dogs: an animal model to study beta-protein
NIH-PA Author Manuscript

amyloidogenesis. In: Maurer, K.; Riederer, P.; Beckman, H., editors. Alzheimer’s disease.
Epidemiology, neuropathology, neurochemistry and clinics. New York: Springer-Verlag; 1990. p.
151-67.
35. Cummings BJ, Su JH, Cotman CW, et al. Beta-amyloid accumulation in aged canine brain: a
model of early plaque formation in Alzheimer’s disease. Neurobiol Aging. 1993; 14(6):547–60.
[PubMed: 8295657]
36. Giaccone G, Verga L, Finazzi M, et al. Cerebral preamyloid deposits and congophilic angiopathy
in aged dogs. Neurosci Lett. 1990; 114:178–83. [PubMed: 2395530]
37. Morys J, Narkiewicz O, Maciejewska B, et al. Amyloid deposits and loss of neurones in the
claustrum of the aged dog. Neuroreport. 1994; 5(14):1825–8. [PubMed: 7827341]
38. Okuda R, Uchida K, Tateyama S, et al. The distribution of amyloid beta precursor protein in canine
brain. Acta Neuropathol. 1994; 87:161–7. [PubMed: 8171967]
39. Russell MJ, White R, Patel E, et al. Familial influence on plaque formation in the beagle brain.
Neuroreport. 1992; 3(12):1093–6. [PubMed: 1493222]
40. Uchida K, Tani Y, Uetsuka K, et al. Immunohistochemical studies on canine cerebral amyloid
angiopathy and senile plaques. J Vet Med Sci. 1992; 54(4):659–67. [PubMed: 1391176]
41. Braak H, Braak E. Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol.
1991; 82(4):239–59. [PubMed: 1759558]
NIH-PA Author Manuscript

42. Braak H, Braak E, Bohl J. Staging of Alzheimer-related cortical destruction. Eur Neurol. 1993;
33:403–8. [PubMed: 8307060]
43. Head E, McCleary R, Hahn FF, et al. Region-specific age at onset of beta-amyloid in dogs.
Neurobiol Aging. 2000; 21(1):89–96. [PubMed: 10794853]
44. Ishihara T, Gondo T, Takahashi M, et al. Immunohistochemical and immunoelectron
microscopical characterization of cerebrovascular and senile plaque amyloid in aged dogs’ brains.
Brain Res. 1991; 548:196–205. [PubMed: 1868335]
45. Thal DR, Rub U, Orantes M, et al. Phases of A beta-deposition in the human brain and its
relevance for the development of AD. Neurology. 2002; 58(12):1791–800. [PubMed: 12084879]
46. Wisniewski HM, Johnson AB, Raine CS, et al. Senile plaques and cerebral amyloidosis in aged
dogs. Lab Invest. 1970; 23:287–96. [PubMed: 5466701]
47. Colle MA, Hauw JJ, Crespeau F, et al. Vascular and parenchymal Ab deposition in the aging dog:
correlation with behavior. Neurobiol Aging. 2000; 21:695–704. [PubMed: 11016539]
48. Cummings BJ, Head E, Afagh AJ, et al. Beta-amyloid accumulation correlates with cognitive
dysfunction in the aged canine. Neurobiol Learn Mem. 1996; 66(1):11–23. [PubMed: 8661247]
49. Head E, Callahan H, Muggenburg BA, et al. Visual-discrimination learning ability and beta-
amyloid accumulation in the dog. Neurobiol Aging. 1998; 19(5):415–25. [PubMed: 9880044]
50. Rofina JE, van Ederen AM, Toussaint MJ, et al. Cognitive disturbances in old dogs suffering from
NIH-PA Author Manuscript

the canine counterpart of Alzheimer’s disease. Brain Res. 2006; 1069(1):216–26. [PubMed:
16423332]
51. Rofina J, van Andel I, van Ederen AM, et al. Canine counterpart of senile dementia of the
Alzheimer type: amyloid plaques near capillaries but lack of spatial relationship with activated
microglia and macrophages. Amyloid. 2003; 10(2):86–96. [PubMed: 12964416]
52. Rofina JE, Singh K, Skoumalova-Vesela A, et al. Histochemical accumulation of oxidative damage
products is associated with Alzheimer-like pathology in the canine. Amyloid. 2004; 11(2):90–100.
[PubMed: 15478464]
53. Selkoe DJ. Soluble oligomers of the amyloid beta-protein impair synaptic plasticity and behavior.
Behav Brain Res. 2008; 192(1):106–13. [PubMed: 18359102]
54. Tomic JL, Pensalfini A, Head E, et al. Soluble fibrillar oligomer levels are elevated in Alzheimer’s
disease brain and correlate with cognitive dysfunction. Neurobiol Dis. 2009; 35(3):352–8.
[PubMed: 19523517]

Vet Clin North Am Small Anim Pract. Author manuscript; available in PMC 2015 November 01.
Vite and Head Page 12

55. Lacor PN, Buniel MC, Chang L, et al. Synaptic targeting by Alzheimer’s-related amyloid beta
oligomers. J Neurosci. 2004; 24(45):10191–200. [PubMed: 15537891]
56. Pop V, Head E, Berchtold NC, et al. Aβ aggregation profiles and shifts in APP processing favor
NIH-PA Author Manuscript

amyloidogenesis in canines. Neurobiol Aging. 2012; 33(1):108–20. [PubMed: 20434811]


57. Attems J. Sporadic cerebral amyloid angiopathy: pathology, clinical implications, and possible
pathomechanisms. Acta Neuropathol. 2005; 110(4):345–59. [PubMed: 16170565]
58. Attems J, Jellinger KA, Lintner F. Alzheimer’s disease pathology influences severity and
topographical distribution of cerebral amyloid angiopathy. Acta Neuropathol. 2005; 110(3):222–
31. [PubMed: 16133541]
59. Herzig MC, Van Nostrand WE, Jucker M. Mechanism of cerebral beta-amyloid angiopathy:
murine and cellular models. Brain Pathol. 2006; 16(1):40–54. [PubMed: 16612981]
60. Prior R, D’Urso D, Frank R, et al. Loss of vessel wall viability in cerebral amyloid angiopathy.
Neuroreport. 1996; 7:562–4. [PubMed: 8730829]
61. Uchida K, Nakayama H, Goto N. Pathological studies on cerebral amyloid angiopathy, senile
plaques and amyloid deposition in visceral organs in aged dogs. J Vet Med Sci. 1991; 53(6):1037–
42. [PubMed: 1790213]
62. Garosi L, McConnell JF, Platt SR, et al. Clinical and topographic magnetic resonance
characteristics of suspected brain infarction in 40 dogs. J Vet Intern Med. 2006; 20(2):311–21.
[PubMed: 16594588]
63. Goncalves R, Carrera I, Garosi L, et al. Clinical and topographic magnetic resonance imaging
characteristics of suspected thalamic infarcts in 16 dogs. Vet J. 2011; 188(1):39–43. [PubMed:
NIH-PA Author Manuscript

20456988]
64. Su MY, Tapp PD, Vu L, et al. A longitudinal study of brain morphometrics using serial magnetic
resonance imaging analysis in a canine model of aging. Prog Neuropsychopharmacol Biol
Psychiatry. 2005; 29(3):389–97. [PubMed: 15795047]
65. Liu S, Hu WX, Zu QQ, et al. A novel embolic stroke model resembling lacunar infarction
following proximal middle cerebral artery occlusion in beagle dogs. J Neurosci Methods. 2012;
209(1):90–6. [PubMed: 22722089]
66. Head E, Moffat K, Das P, et al. Beta-amyloid deposition and tau phosphorylation in clinically
characterized aged cats. Neurobiol Aging. 2005; 26(5):749–63. [PubMed: 15708450]
67. Yu CH, Song GS, Yhee JY, et al. Histopathological and immunohistochemical comparison of the
brain of human patients with Alzheimer’s disease and the brain of aged dogs with cognitive
dysfunction. J Comp Pathol. 2011; 145(1):45–58. [PubMed: 21256508]
68. Head E, Liu J, Hagen TM, et al. Oxidative damage increases with age in a canine model of human
brain aging. J Neurochem. 2002; 82:375–81. [PubMed: 12124438]
69. Skoumalova A, Rofina J, Schwippelova Z, et al. The role of free radicals in canine counterpart of
senile dementia of the Alzheimer type. Exp Gerontol. 2003; 38:711–9. [PubMed: 12814808]
70. Kiatipattanasakul W, Nakamura S, Kuroki K, et al. Immunohistochemical detection of anti-
oxidative stress enzymes in the dog brain. Neuropathology. 1997; 17:307–12.
NIH-PA Author Manuscript

71. Opii WO, Joshi G, Head E, et al. Proteomic identification of brain proteins in the canine model of
human aging following a long-term treatment with antioxidants and a program of behavioral
enrichment: relevance to Alzheimer’s disease. Neurobiol Aging. 2008; 29(1):51–70. [PubMed:
17055614]
72. Hwang IK, Yoon YS, Yoo KY, et al. Differences in lipid peroxidation and Cu,Zn-superoxide
dismutase in the hippocampal CA1 region between adult and aged dogs. J Vet Med Sci. 2008;
70(3):273–7. [PubMed: 18388427]
73. Papaioannou N, Tooten PC, van Ederen AM, et al. Immunohistochemical investigation of the brain
of aged dogs. I. Detection of neurofibrillary tangles and of 4-hydroxynonenal protein, an oxidative
damage product, in senile plaques. Amyloid. 2001; 8:11–21. [PubMed: 11293821]
74. Shigenaga MK, Hagen TM, Ames BN. Oxidative damage and mitochondrial decay in aging. Proc
Natl Acad Sci U S A. 1994; 91:10771–8. [PubMed: 7971961]
75. Head E, Nukala VN, Fenoglio KA, et al. Effects of age, dietary, and behavioral enrichment on
brain mitochondria in a canine model of human aging. Exp Neurol. 2009; 220(1):171–6. [PubMed:
19703441]

Vet Clin North Am Small Anim Pract. Author manuscript; available in PMC 2015 November 01.
Vite and Head Page 13

76. Kuruppu DK, Matthews BR. Young-onset dementia. Semin Neurol. 2013; 33(4):365–85.
[PubMed: 24234358]
77. Swain GP, Prociuk M, Bagel JH, et al. Adeno-associated virus serotypes 9 and rh10 mediate strong
NIH-PA Author Manuscript

neuronal transduction of the dog brain. Gene Ther. 2014; 21(1):28–36. [PubMed: 24131981]
78. Ellinwood NM, Vite CH, Haskins ME. Gene therapy for lysosomal storage diseases: the lessons
and promise of animal models. J Gene Med. 2004; 6(5):481–506. [PubMed: 15133760]
79. Raz N, Rodrigue KM, Haacke EM. Brain aging and its modifiers: insights from in vivo
neuromorphometry and susceptibility weighted imaging. Ann N Y Acad Sci. 2007; 1097:84–93.
[PubMed: 17413014]
80. Dolisca SB, Mehta M, Pearce DA, et al. Batten disease: clinical aspects, molecular mechanisms,
translational science, and future directions. J Child Neurol. 2013; 28(9):1074–100. [PubMed:
23838031]
81. Hofmann, SL.; Peltonen, L. The Neuronal Ceroid Lipofuscinoses. In: Valle, D.; Beaudet, AL.;
Vogelstein, B., et al., editors. OMMBID - The Online Metabolic and Molecular Bases of Inherited
Diseases. New York, NY: McGraw-Hill; 2014. http://ommbid.mhmedical.com/content.aspx?
bookid=474&Sectionid=45374157 [Accessed August 20, 2014]
82. Jolly RD. Comparative biology of the neuronal ceroid-lipofuscinoses (NCL): an overview. Am J
Med Genet. 1995; 57(2):307–11. [PubMed: 7668352]
83. Jolly RD, Brown S, Das AM, et al. Mitochondrial dysfunction in the neuronal ceroid-
lipofuscinoses (Batten disease). Neurochem Int. 2002; 40(6):565–71. [PubMed: 11850114]
84. Sanders DN, Kanazono S, Wininger FA, et al. A reversal learning task detects cognitive deficits in
NIH-PA Author Manuscript

a Dachshund model of late-infantile neuronal ceroid lipofuscinosis. Genes Brain Behav. 2011;
10(7):798–804. [PubMed: 21745338]
85. Vuillemenot BR, Katz ML, Coates JR, et al. Intrathecal tripeptidyl-peptidase 1 reduces lysosomal
storage in a canine model of late infantile neuronal ceroid lipofuscinosis. Mol Genet Metab. 2011;
104(3):325–37. [PubMed: 21784683]
86. Shapiro E, Guler OE, Rudser K, et al. An exploratory study of brain function and structure in
mucopolysaccharidosis type I: long term observations following hematopoietic cell transplantation
(HCT). Mol Genet Metab. 2012; 107(1–2):116–21. [PubMed: 22867884]
87. Vite CH, Nestrasil I, Mlikotic A, et al. Features of brain MRI in dogs with treated and untreated
mucopolysaccharidosis type I. Comp Med. 2013; 63(2):163–73. [PubMed: 23582423]
88. Pelissier C, Roudier M, Boller F. Factorial validation of the severe impairment battery for patients
with Alzheimer’s disease. A pilot study. Dement Geriatr Cogn Disord. 2002; 13(2):95–100.
[PubMed: 11844891]
89. Ferris S, Karantzoulis S, Somogyi M, et al. Rivastigmine in moderately severe-to-severe
Alzheimer’s disease: severe impairment battery factor analysis. Alzheimers Res Ther. 2013; 5(6):
63. [PubMed: 24351447]
90. Dierenfeld AD, McEntee MF, Vogler CA, et al. Replacing the enzyme alpha-L-iduronidase at birth
ameliorates symptoms in the brain and periphery of dogs with mucopolysaccharidosis type I. Sci
Transl Med. 2010; 2(60):60ra89.
NIH-PA Author Manuscript

91. Chen A, Vogler C, McEntee M, et al. Glycosaminoglycan storage in neuroanatomical regions of


mucopolysaccharidosis I dogs following intrathecal recombinant human iduronidase. APMIS.
2011; 119(8):513–21. [PubMed: 21749451]
92. Gunn-Moore D, Moffat K, Christie LA, et al. Cognitive dysfunction and the neurobiology of
ageing in cats. J Small Anim Pract. 2007; 48(10):546–53. [PubMed: 17617164]
93. Landsberg GM, Denenberg S, Araujo JA. Cognitive dysfunction in cats: a syndrome we used to
dismiss as ‘old age’. J Feline Med Surg. 2010; 12(11):837–48. [PubMed: 20974401]
94. Levine MS, Lloyd RL, Fisher RS, et al. Sensory, motor and cognitive alterations in aged cats.
Neurobiol Aging. 1987; 8:253–63. [PubMed: 3600956]
95. Levine MS, Adinolfi AM, Fisher RS, et al. Quantitative morphology of medium-sized caudate
spiny neurons in aged cats. Neurobiol Aging. 1986; 7(4):277–86. [PubMed: 3748270]
96. Levine MS, Adinolfi AM, Fisher RS, et al. Ultrastructural alterations in caudate nucleus in aged
cats. Brain Res. 1988; 440(2):267–79. [PubMed: 3359214]

Vet Clin North Am Small Anim Pract. Author manuscript; available in PMC 2015 November 01.
Vite and Head Page 14

97. Levine MS. Neurophysiological and morphological alterations in caudate neurons in aged cats.
Ann N Y Acad Sci. 1988; 515:314–28. [PubMed: 3364891]
98. Zhang JH, Sampogna S, Morales FR, et al. Age-related changes in cholinergic neurons in the
NIH-PA Author Manuscript

laterodorsal and the pedunculo-pontine tegmental nuclei of cats: a combined light and electron
microscopic study. Brain Res. 2005; 1052(1):47–55. [PubMed: 16002054]
99. Cummings BJ, Satou T, Head E, et al. Diffuse plaques contain C-terminal A beta 42 and not A beta
40: evidence from cats and dogs. Neurobiol Aging. 1996; 17(4):653–9. [PubMed: 8832640]
100. Gunn-Moore DA, McVee J, Bradshaw JM, et al. Ageing changes in cat brains demonstrated by
beta-amyloid and AT8-immunoreactive phosphorylated tau deposits. J Feline Med Surg. 2006;
8(4):234–42. [PubMed: 16603401]
101. Nakamura S, Nakayama H, Kiatipattanasakul W, et al. Senile plaques in very aged cats. Acta
Neuropathol. 1996; 91:437–9. [PubMed: 8928623]
102. Brellou G, Vlemmas I, Lekkas S, et al. Immunohistochemical investigation of amyloid beta-
protein (Abeta) in the brain of aged cats. Histol Histopathol. 2005; 20(3):725–31. [PubMed:
15944921]
103. Takeuchi Y, Uetsuka K, Murayama M, et al. Complementary distributions of amyloid-beta and
neprilysin in the brains of dogs and cats. Vet Pathol. 2008; 45(4):455–66. [PubMed: 18587091]
104. Chambers JK, Mutsuga M, Uchida K, et al. Characterization of AbetapN3 deposition in the brains
of dogs of various ages and other animal species. Amyloid. 2011; 18(2):63–71. [PubMed:
21557687]
105. Goedert M, Jakes R, Crowther RA, et al. The abnormal phosphorylation of tau protein at Ser-202
NIH-PA Author Manuscript

in Alzheimer disease recapitulates phosphorylation during development. Proc Natl Acad Sci U S
A. 1993; 90:5066–70. [PubMed: 8506352]
106. Braak H, Braak E. Staging of Alzheimer’s disease-related neurofibrillary changes. Neurobiol
Aging. 1995; 16(3):271–84. [PubMed: 7566337]
107. Janke C, Beck M, Stahl T, et al. Phylogenetic diversity of the expression of the microtubule-
associated protein tau: implications for neurodegenerative disorders. Brain Res Mol Brain Res.
1999; 68:119–28. [PubMed: 10320789]
108. Braak H, Braak E, Strothjohann M. Abnormally phosphorylated tau protein related to the
formation of neurofibrillary tangles and neuropil threads in the cerebral cortex of sheep and goat.
Neurosci Lett. 1994; 171:1–4. [PubMed: 7521944]
109. Kuroki K, Uchida K, Kiatipattanasakul W, et al. Immunohistochemical detection of tau proteins
in various non-human animal brains. Neuropathology. 1997; 17:174–80.
110. Walkley SU, Suzuki K. Consequences of NPC1 and NPC2 loss of function in mammalian
neurons. Biochim Biophys Acta. 2004; 1685(1–3):48–62. [PubMed: 15465426]
111. March PA, Thrall MA, Brown DE, et al. GABAergic neuroaxonal dystrophy and other
cytopathological alterations in feline Niemann-Pick disease type C. Acta Neuropathol. 1997;
94(2):164–72. [PubMed: 9255392]
112. Zervas M, Somers KL, Thrall MA, et al. Critical role for glycosphingolipids in Niemann-Pick
NIH-PA Author Manuscript

disease type C. Curr Biol. 2001; 11(16):1283–7. [PubMed: 11525744]


113. Somers KL, Brown DE, Fulton R, et al. Effects of dietary cholesterol restriction in a feline model
of Niemann-Pick type C disease. J Inherit Metab Dis. 2001; 24(4):427–36. [PubMed: 11596647]
114. Ward S, O’Donnell P, Fernandez S, et al. 2-Hydroxypropyl-beta-cyclodextrin raises hearing
threshold in normal cats and in cats with Niemann-Pick type C disease. Pediatr Res. 2010; 68(1):
52–6. [PubMed: 20357695]
115. Ferris S, Cummings J, Christensen D, et al. Effects of donepezil 23 mg on severe impairment
battery domains in patients with moderate to severe Alzheimer’s disease: evaluating the impact
of baseline severity. Alzheimers Res Ther. 2013; 5(1):12. [PubMed: 23433097]
116. Sarkar S, Carroll B, Buganim Y, et al. Impaired autophagy in the lipid-storage disorder Niemann-
pick type C1 disease. Cell Rep. 2013; 5(5):1302–15. [PubMed: 24290752]
117. Hara T, Nakamura K, Matsui M, et al. Suppression of basal autophagy in neural cells causes
neurodegenerative disease in mice. Nature. 2006; 441(7095):885–9. [PubMed: 16625204]
118. Keller JN, Dimayuga E, Chen Q, et al. Autophagy, proteasomes, lipofuscin, and oxidative stress
in the aging brain. Int J Biochem Cell Biol. 2004; 36(12):2376–91. [PubMed: 15325579]

Vet Clin North Am Small Anim Pract. Author manuscript; available in PMC 2015 November 01.
Vite and Head Page 15

119. Komatsu M, Waguri S, Chiba T, et al. Loss of autophagy in the central nervous system causes
neurodegeneration in mice. Nature. 2006; 441(7095):880–4. [PubMed: 16625205]
120. Kiselyov K, Jennigs JJ Jr, Rbaibi Y, et al. Autophagy, mitochondria and cell death in lysosomal
NIH-PA Author Manuscript

storage diseases. Autophagy. 2007; 3(3):259–62. [PubMed: 17329960]


121. Fu R, Yanjanin NM, Bianconi S, et al. Oxidative stress in Niemann-Pick disease, type C. Mol
Genet Metab. 2010; 101(2–3):214–8. [PubMed: 20667755]
122. Porter FD, Scherrer DE, Lanier MH, et al. Cholesterol oxidation products are sensitive and
specific blood-based biomarkers for Niemann-Pick C1 disease. Sci Transl Med. 2010; 2(56):
56ra81.
123. Fu R, Yanjanin NM, Elrick MJ, et al. Apolipoprotein E genotype and neurological disease onset
in Niemann-Pick disease, type C1. Am J Med Genet A. 2012; 158A(11):2775–80. [PubMed:
23023945]
124. Mattsson N, Olsson M, Gustavsson MK, et al. Amyloid-beta metabolism in Niemann-Pick C
disease models and patients. Metab BrainDis. 2012; 27(4):573–85.
125. Schedin S, Sindelar PJ, Pentchev P, et al. Peroxisomal impairment in Niemann-Pick type C
disease. J Biol Chem. 1997; 272(10):6245–51. [PubMed: 9045641]
126. Lloyd-Evans E, Morgan AJ, He X, et al. Niemann-Pick disease type C1 is a sphingosine storage
disease that causes deregulation of lysosomal calcium. Nat Med. 2008; 14(11):1247–55.
[PubMed: 18953351]
127. Baudry M, Yao Y, Simmons D, et al. Postnatal development of inflammation in a murine model
of Niemann-Pick type C disease: immunohistochemical observations of microglia and astroglia.
NIH-PA Author Manuscript

Exp Neurol. 2003; 184(2):887–903. [PubMed: 14769381]


128. Wu YP, Mizukami H, Matsuda J, et al. Apoptosis accompanied by up-regulation of TNF-alpha
death pathway genes in the brain of Niemann-Pick type C disease. Mol Genet Metab. 2005;
84(1):9–17. [PubMed: 15639190]
129. van Echten-Deckert G, Walter J. Sphingolipids: critical players in Alzheimer’s disease. Prog
Lipid Res. 2012; 51(4):378–93. [PubMed: 22835784]
130. Houpt KA, Beaver B. Behavioral problems of geriatric dogs and cats. Vet Clin North Am Small
Anim Pract. 1981; 11(4):643–52. [PubMed: 6977925]
131. Mosier JE. Effect of aging on body systems of the dog. Vet Clin North Am Small Anim Pract.
1989; 19(1):1–12. [PubMed: 2646811]
132. Chapman BL, Voith VL. Behavioral problems in old dogs: 26 cases (1984–1987). J Am Vet Med
Assoc. 1990; 196(6):944–6. [PubMed: 2312394]
133. Ruehl WW, Bruyette DS, DePaoli A, et al. Canine cognitive dysfunction as a model for human
age-related cognitive decline, dementia and Alzheimer’s disease: clinical presentation, cognitive
testing, pathology and response to 1-deprenyl therapy. Prog Brain Res. 1995; 106:217–25.
[PubMed: 8584657]
134. Landsberg GM, Nichol J, Araujo JA. Cognitive dysfunction syndrome: a disease of canine and
feline brain aging. Vet Clin North Am Small Anim Pract. 2012; 42(4):749–68. vii. [PubMed:
NIH-PA Author Manuscript

22720812]
135. Bosch MN, Pugliese M, Gimeno-Bayon J, et al. Dogs with cognitive dysfunction syndrome: a
natural model of Alzheimer’s disease. Curr Alzheimer Res. 2012; 9(3):298–314. [PubMed:
21875411]
136. Pugliese M, Geloso MC, Carrasco JL, et al. Canine cognitive deficit correlates with diffuse
plaque maturation and S100beta (−) astrocytosis but not with insulin cerebrospinal fluid level.
Acta Neuropathol. 2006; 111(6):519–28. [PubMed: 16718348]
137. Pugliese M, Mascort J, Mahy N, et al. Diffuse beta-amyloid plaques and hyper-phosphorylated
tau are unrelated processes in aged dogs with behavioral deficits. Acta Neuropathol. 2006;
112(2):175–83. [PubMed: 16775693]
138. Head E, Cotman CW, Zicker SC, et al. The use of dietary antioxidants and mitochondrial co-
factors to promote successful aging. Curr Top Nutraceutical Res. 2005; 3(2):85–94.
139. Bosch MN, Gimeno-Bayon J, Rodriguez MJ, et al. Rapid improvement of canine cognitive
dysfunction with immunotherapy designed for Alzheimer’s disease. Curr Alzheimer Res. 2013;
10(5):482–93. [PubMed: 23566345]

Vet Clin North Am Small Anim Pract. Author manuscript; available in PMC 2015 November 01.
Vite and Head Page 16

140. Gunn-Moore DA. Cognitive dysfunction in cats: clinical assessment and management. Top
Companion Anim Med. 2011; 26(1):17–24. [PubMed: 21435622]
NIH-PA Author Manuscript
NIH-PA Author Manuscript
NIH-PA Author Manuscript

Vet Clin North Am Small Anim Pract. Author manuscript; available in PMC 2015 November 01.
Vite and Head Page 17

KEY POINTS
• Brain atrophy, neuron loss, decreased neurogenesis, and oxidative stress but few
NIH-PA Author Manuscript

tau-associated disorders are observed in aging dog brains.

• Cerebrovascular pathology can be extensive in canine brain aging.

• β-Amyloid protein, associated with Alzheimer disease in humans, is increased


with age in the dog brain and is linked to signs of learning and memory
impairments.

• Lysosomal storage diseases in dogs are associated with similar types of


neuropathology as are observed with aging and Alzheimer disease.

• Few studies describe the neurobiology of aging in cats but interesting


similarities and differences from dogs have been reported.

• Feline Niemann-Pick type C disease has several neuropathologic and clinical


similarities to Alzheimer disease.
NIH-PA Author Manuscript
NIH-PA Author Manuscript

Vet Clin North Am Small Anim Pract. Author manuscript; available in PMC 2015 November 01.
Vite and Head Page 18
NIH-PA Author Manuscript

Fig. 1.
Aβ immunostaining (brown) in the brain of a 14-year-old border collie (Martha) with signs
of cognitive dysfunction syndrome. Aβ was detected using the 6E10 antibody that binds Aβ
NIH-PA Author Manuscript

1 to 16. (A) Low-power magnification (1.5×) of extensive Aβ deposition in the


hippocampus. (B) Aβ is primarily found in the outer molecular layer of the dentate gyrus,
which contains neuron terminals originating from the entorhinal cortex (4×). (C) The
prefrontal cortex also shows extensive Aβ deposition that appears most dense in layers II
and V of the cortex and is less apparent in the white matter (1.5×). (D) At high
magnification (4×), the differential deposition of Aβ in the 6 cortical layers can be seen as
well as extensive white matter cerebral amyloid angiopathy (arrow). Sections have been
counterstained with cresyl violet (blue).
NIH-PA Author Manuscript

Vet Clin North Am Small Anim Pract. Author manuscript; available in PMC 2015 November 01.
Vite and Head Page 19
NIH-PA Author Manuscript
NIH-PA Author Manuscript

Fig. 2.
Aging and canine cognitive dysfunction in a border collie (Martha). (A) Martha competing
in sheep dog trials when she was young, and (B) still active around the age of 10 years. As
she reached 11 years of age she struggled to complete commands issued by her owner (C).
At 14 years of age, Martha was generally in good health except for arthritis but would not
make eye contact with her owner (D).
NIH-PA Author Manuscript

Vet Clin North Am Small Anim Pract. Author manuscript; available in PMC 2015 November 01.
Vite and Head Page 20

Table 1

Comparison of dog and cat neurologic neurodegenerative features


NIH-PA Author Manuscript

Neurobiological Outcome Measures Canine Feline NCL or MPSI NPC Disease


Brain atrophy Yes NA Yes Yes

β-Amyloid Yes Yes Yes Yes

Tau Yes Yes NA Yes

Cerebral amyloid angiopathy Yes Yes NA NA

Infarcts Yes NA NA NA

Vascular disease Yes NA Yes NA

Lipofuscin accumulation Yes Yes Yes NA

Caspase activation Yes NA Yes Yes

DNA fragmentation Yes NA NA NA

Neuron loss: hippocampus Yes NA Yes Yes

Neuron loss: caudate NA Yes Yes Yes

Neuron loss: locus coeruleus Yes Yes NA NA

Neuron loss: Purkinje cerebellar cells Yes NA Yes Yes


NIH-PA Author Manuscript

White matter degeneration Yes NA Yes Yes

Inflammation Yes NA Yes Yes

Oxidative damage Yes NA Yes Yes

Gliosis Yes NA Yes Yes

Abbreviations: MPSI, mucopolysaccharidosis type I; NA, not available; NCL, neuronal ceroid lipofuscinoses; NPC, Niemann-Pick type C.
NIH-PA Author Manuscript

Vet Clin North Am Small Anim Pract. Author manuscript; available in PMC 2015 November 01.

You might also like